Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 276
Filtrar
1.
Mol Imaging Biol ; 25(2): 384-390, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35999424

RESUMEN

PURPOSE: 6-O-(2-[18F]Fluoroethyl)-6-O-desmethyl-diprenorphine ([18F]FE-DPN) is regarded as a non-selective opioid receptor radiotracer. PROCEDURE: Here, we report the first characterization of [18F]FE-DPN synthesized from the novel precursor, 6-O-(2-tosyloxyethoxy)-6-O-desmethyl-3-O-trityl-diprenorphine (TE-TDDPN), using a one-pot, two-step nucleophilic radiosynthesis to image opioid receptors in rats and mice using positron emission tomography. RESULTS: We also show that [18F]FE-DPN and [3H]DPN exhibit negligible brain uptake in mu opioid receptor (MOR) knockout mice. CONCLUSIONS: Taken together with prior findings, our results suggest that [18F]FE-DPN and [3H]DPN preferentially bind to MOR in rodents in vivo.


Asunto(s)
Tomografía de Emisión de Positrones , Receptores Opioides mu , Ratas , Ratones , Animales , Diprenorfina/metabolismo , Receptores Opioides mu/metabolismo , Tomografía de Emisión de Positrones/métodos , Encéfalo/metabolismo , Receptores Opioides/metabolismo
2.
Hum Brain Mapp ; 31(4): 550-5, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19780041

RESUMEN

Animal and functional imaging studies had identified cortical structures such as the parieto-insular vestibular cortex, the retro-insular cortex, or the anterior cingulate cortex belonging to a vestibular cortical network. Basic animal studies revealed that endorphins might be important transmitters involved in cerebral vestibular processing. The aim of the present study was therefore to analyse whether the opioid system is involved in vestibular neurotransmission of humans or not. Changes in opioid receptor availability during caloric air stimulation of the right ear were studied with [(18)F] Fluoroethyl-diprenorphine ([(18)F]FEDPN) PET scans in 10 right-handed healthy volunteers and compared to a control condition. Decrease in receptor availability to [(18)F]FEDPN during vestibular stimulation in comparison to the control condition was significant at the right posterior insular cortex and the postcentral region indicating more endogenous opioidergic binding in these regions during stimulation. These data give evidence that the opioidergic system plays a role in the right hemispheric dominance of the vestibular cortical system in right-handers.


Asunto(s)
Encéfalo/metabolismo , Mareo/metabolismo , Receptores Opioides/metabolismo , Vértigo/metabolismo , Vestíbulo del Laberinto/fisiología , Adulto , Presión Sanguínea , Encéfalo/diagnóstico por imagen , Diprenorfina/análogos & derivados , Diprenorfina/metabolismo , Mareo/diagnóstico por imagen , Humanos , Masculino , Estimulación Física , Tomografía de Emisión de Positrones , Transmisión Sináptica/fisiología , Vértigo/diagnóstico por imagen
3.
J Pharmacol Exp Ther ; 335(3): 562-71, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20829393

RESUMEN

Buprenorphine is a partial µ-opioid receptor agonist used for the treatment of opioid dependence that has several advantages over methadone. The principal route of buprenorphine disposition has been well established; however, little is known regarding the potential for buprenorphine to influence the metabolism and clearance of other drugs by affecting the expression of drug-metabolizing enzymes (DMEs). Here, we investigate the effects of buprenorphine on the activation of pregnane X receptor (PXR) and constitutive androstane receptor (CAR), as well as the induction of DMEs, in both HepG2 cells and human primary hepatocytes (HPHs). In HepG2 cells, buprenorphine significantly increased human PXR-mediated CYP2B6 and CYP3A4 reporter activities. CYP2B6 reporter activity was also enhanced by buprenorphine in HepG2 cells cotransfected with a chemical-responsive human CAR variant. Real-time reverse transcription-polymerase chain reaction analysis revealed that buprenorphine strongly induced CYP3A4 expression in both PXR- and CAR-transfected HepG2 cells. However, treatment with the same concentrations of buprenorphine in HPHs resulted in literally no induction of CYP3A4 or CYP2B6 expression. Further studies indicated that buprenorphine could neither translocate human CAR to the nucleus nor activate CYP2B6/CYP3A4 reporter activities in transfected HPHs. Subsequent experiments to determine whether the differential response was due to buprenorphine's metabolic stability revealed a dramatically differential rate of elimination for buprenorphine between HPHs and HepG2 cells. Taken together, these studies indicate that metabolic stability of buprenorphine defines the differential induction of DMEs observed in HepG2 and HPHs, and the results obtained from PXR and CAR reporter assays in immortalized cell line require cautious interpretation.


Asunto(s)
Buprenorfina/metabolismo , Buprenorfina/farmacología , Hepatocitos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Analgésicos Opioides/metabolismo , Hidrocarburo de Aril Hidroxilasas/genética , Hidrocarburo de Aril Hidroxilasas/metabolismo , Receptor de Androstano Constitutivo , Citocromo P-450 CYP2B6 , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Diprenorfina/metabolismo , Diprenorfina/farmacología , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Genes Reporteros/genética , Células Hep G2 , Hepatocitos/efectos de los fármacos , Humanos , Oxidorreductasas N-Desmetilantes/genética , Oxidorreductasas N-Desmetilantes/metabolismo , Receptor X de Pregnano , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Esteroides/agonistas , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/genética , Transfección
4.
Neuroscience ; 159(1): 405-13, 2009 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-19138727

RESUMEN

Prenatal morphine treatment and emotional stress both have been shown to increase sensitivity to reward-related behaviors. It has been postulated that this increased sensitivity to rewarding stimuli may be the result of an enhanced release of endogenous opioids. In the present study, in vivo autoradiography was employed to investigate the endogenous opioid release in specific brain areas in rats. Pregnant animals were exposed to morphine or saline from day 8 of gestation till birth. Development of pups was monitored and play behavior was tested on postnatal day 21. Adult rats were exposed to repeated emotional stress or control treatment for five consecutive days and tested in a small open field 5 days later. [(3)H]-Diprenorphine was injected following this test to investigate endogenous opioid release. Prenatal morphine treatment increased play behavior and endogenous opioid release in a number of cortical and subcortical brain areas after being subjected to an open field challenge later in life. Emotional stress exposure increased locomotor activity in the open field irrespective of the type of prenatal treatment and increased endogenous opioid release in some specific brain areas. It is suggested that the increased release of endogenous opioids in the substantia nigra, the piriform cortex and the septum observed after both types of treatments is related to the increased sensitivity to reward.


Asunto(s)
Analgésicos Opioides/metabolismo , Encéfalo/metabolismo , Morfina/administración & dosificación , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Estrés Psicológico , Análisis de Varianza , Animales , Animales Recién Nacidos , Autorradiografía/métodos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Diprenorfina/administración & dosificación , Diprenorfina/metabolismo , Conducta Exploratoria/fisiología , Femenino , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/metabolismo , Embarazo , Ratas , Ratas Wistar , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Estrés Psicológico/fisiopatología , Tritio/metabolismo
5.
Science ; 258(5090): 1952-5, 1992 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-1335167

RESUMEN

Opiate drugs have potent analgesic and addictive properties. These drugs interact with receptors that also mediate the response to endogenous opioid peptide ligands. However, the receptors for opioids have eluded definitive molecular characterization. By transient expression in COS cells and screening with an iodinated analog of the opioid peptide enkephalin, a complementary DNA clone encoding a functional delta opioid receptor has been identified. The sequence shows homology to G protein-coupled receptors, in particular the receptors for somatostatin, angiotensin, and interleukin-8.


Asunto(s)
Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Secuencia de Aminoácidos , Animales , Unión Competitiva , Northern Blotting , Southern Blotting , Línea Celular , AMP Cíclico/metabolismo , Diprenorfina/metabolismo , Encefalina D-Penicilamina (2,5) , Encefalinas/farmacología , Etorfina/farmacología , Expresión Génica , Humanos , Cinética , Modelos Estructurales , Datos de Secuencia Molecular , Naloxona/farmacología , Narcóticos/farmacología , Estructura Secundaria de Proteína , Receptores Opioides delta/química , Homología de Secuencia de Aminoácido , Transfección , Células Tumorales Cultivadas
6.
Science ; 221(4616): 1198-201, 1983 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-6310750

RESUMEN

In vitro binding studies have demonstrated the existence of multiple opiate receptor types. An additional site in the rat brain (termed the lambda site) is distinct from the established types by its selectivity for 4,5-epoxymorphinans (such as naloxone and morphine). While the lambda site displays a high affinity for naloxone in vivo and in vitro in fresh brain membrane homogenates, these sites rapidly convert in vitro to a state of low affinity. The regional distribution of the lambda site in the brain is strikingly different from that of the classic opiate receptor types.


Asunto(s)
Química Encefálica , Naloxona/metabolismo , Receptores Opioides/metabolismo , Animales , Dihidromorfina/metabolismo , Diprenorfina/metabolismo , Morfina/metabolismo , Nalorfina/metabolismo , Naltrexona/metabolismo , Ratas , Sodio/metabolismo , Distribución Tisular
7.
Biochem J ; 412(2): 245-56, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18307412

RESUMEN

Opioid agonists have a broad range of effects on cells of the immune system, including modulation of the inflammatory response, and opioid and chemokine receptors are co-expressed by many white cells. Hetero-oligomerization of the human DOP opioid and chemokine CXCR2 receptors could be detected following their co-expression by each of co-immunoprecipitation, three different resonance energy transfer techniques and the construction of pairs of individually inactive but potentially complementary receptor G-protein alpha subunit fusion proteins. Although DOP receptor agonists and a CXCR2 antagonist had no inherent affinity for the alternative receptor when either receptor was expressed individually, use of cells that expressed a DOP opioid receptor construct constitutively, and in which expression of a CXCR2 receptor construct could be regulated, demonstrated that the CXCR2 antagonist enhanced the function of DOP receptor agonists only in the presence of CXCR2. This effect was observed for both enkephalin- and alkaloid-based opioid agonists, and the effective concentrations of the CXCR2 antagonist reflected CXCR2 receptor occupancy. Entirely equivalent results were obtained in cells in which the native DOP opioid receptor was expressed constitutively and in which expression of the isolated CXCR2 receptor could be induced. These results indicate that a CXCR2 receptor antagonist can enhance the function of agonists at a receptor for which it has no inherent direct affinity by acting as an allosteric regulator of a receptor that is a heterodimer partner for the CXCR2 receptor. These results have novel and important implications for the development and use of small-molecule therapeutics.


Asunto(s)
Estructura Cuaternaria de Proteína , Receptores de Interleucina-8B , Receptores Opioides delta/química , Receptores Opioides delta/metabolismo , Regulación Alostérica , Línea Celular , Dimerización , Diprenorfina/química , Diprenorfina/metabolismo , Leucina Encefalina-2-Alanina/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Humanos , Antagonistas de Narcóticos/química , Antagonistas de Narcóticos/metabolismo , Compuestos de Fenilurea/metabolismo , Receptores de Interleucina-8B/antagonistas & inhibidores , Receptores de Interleucina-8B/química , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo , Receptores Opioides delta/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
8.
Biochemistry ; 47(40): 10576-86, 2008 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-18778083

RESUMEN

We previously demonstrated that D3.49(164)Y or T6.34(279)K mutation in the rat mu opioid receptor (MOPR) resulted in agonist-independent activation. Here, we identified the cysteine(s) within the transmembrane domains (TMs) of the D3.49(164)Y mutant that became accessible in the binding-site crevice by use of methanethiosulfonate ethylammonium (MTSEA) and inferred conformational changes associated with receptor activation. While the C7.38(321)S mutant was insensitive to MTSEA, the D3.49(164)Y/C7.38(321)S mutant showed similar sensitivity as the D3.49(164)Y, suggesting that, in the D3.49(164)Y mutant, C7.38(321) becomes inaccessible while other cysteines are accessible in the binding-site crevice. Each of the other seven cysteines in the TMs was mutated to serine on the background of D3.49(164)Y/C7.38(321)S, and the resulting triple mutants were evaluated for [3H]diprenorphine and [d-Ala2,NMe-Phe4,Gly5-ol]-enkephalin (DAMGO) binding and effect of MTSEA on [3H]diprenorphine binding. The D3.49(164)Y/C7.38(321)S mutant and the triple mutants, except the C6.47(292)S triple mutant, retained similar affinities for [3H]diprenorphine and DAMGO as the D3.49(164)Y mutant. The second-order rate constants for MTSEA reactions showed that C3.44(159)S, C4.48(190)S, C5.41(235)S, and C7.47(330)S significantly reduced sensitivity to MTSEA, compared with the D3.49(164)Y/C7.38(321)S. These results suggest that the four cysteines may be rotated and/or tilted to become accessible. While the D3.49(164)Y/C7.38(321)S was similarly sensitive to MTSEA as the D3.49(164)Y mutant, the T6.34(279)K/C7.38(321)S was much less sensitive to MTSEA than the T6.34(279)K mutant, suggesting that the two constitutively active mutants assume different conformations and/or possess different dynamic properties. Molecular models of the MOPR monomer and homodimer, using the crystal structures of rhodopsin, the beta2-adrenergic receptor, and the ligand-free opsin, which contains several features characteristic of the active state, were employed to analyze these experimental results in a structural context.


Asunto(s)
Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Diprenorfina/metabolismo , Diprenorfina/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Terciaria de Proteína/genética , Ratas , Receptores Opioides mu/genética
9.
J Nucl Med ; 49(8): 1257-61, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18632824

RESUMEN

UNLABELLED: Little is known about the neurobiologic correlates of human personality. On the basis of the key role of the central opioidergic system in addiction and substance abuse, we investigated the relationship between certain personality traits that are supposed to be relevant in addiction and the opioid receptor status in healthy subjects. METHODS: We investigated 23 healthy male volunteers who were extensively clinically tested to exclude substance abuse. All of the subjects underwent 1 PET scan with the subtype-nonselective opioidergic radioligand 18F-fluoroethyl-diprenorphine under resting conditions without sensory or cognitive stimulation. Subsequently, the subjects were psychologically tested for the personality traits novelty seeking, harm avoidance, reward dependence, and persistence, according to Cloninger's biosocial model of personality. The binding potential (BP) as a parameter of regional cerebral opioid receptor availability was computed by means of the modified Logan plot using the occipital cortex as a reference region. Further imaging data analysis was performed using statistical parametric mapping; after stereotactic normalization, the correlations were calculated between the regional BP and the psychologic scores on a voxel-by-voxel basis. RESULTS: The correlation analysis between personality dimensions and opioid receptor availability showed a significant (P < 0.001) positive correlation between the scores of reward dependence and the BP of the bilateral ventral striatum with nucleus accumbens (z scores, 4.52 and 4.33, respectively). The additionally performed region-of-interest-based correlation analysis yielded correlation coefficients of r = 0.84 and r = 0.81 for the left and right ventral striata, respectively. No further significant correlations were detectable between the other personality dimensions and cerebral opioid receptor binding. CONCLUSION: In healthy subjects, personality traits, which might be predisposing for addictive behavior, are correlated to the opioidergic neurotransmission in core structures of the human reward system.


Asunto(s)
Encéfalo/metabolismo , Diprenorfina/análogos & derivados , Radiofármacos , Receptores Opioides/metabolismo , Recompensa , Adulto , Ganglios Basales/diagnóstico por imagen , Ganglios Basales/metabolismo , Encéfalo/diagnóstico por imagen , Mapeo Encefálico , Núcleo Caudado/diagnóstico por imagen , Núcleo Caudado/metabolismo , Diprenorfina/metabolismo , Radioisótopos de Flúor , Humanos , Masculino , Persona de Mediana Edad , Núcleo Accumbens/diagnóstico por imagen , Núcleo Accumbens/metabolismo , Personalidad , Cintigrafía , Radiofármacos/metabolismo
10.
Bioorg Med Chem Lett ; 18(6): 2006-12, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18313920
11.
Peptides ; 28(12): 2340-7, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18022288

RESUMEN

Met-enkephalin-Gly-Tyr (MEGY) is an endogenous peptide that binds to opioid sites in zebrafish and in rat brain homogenates. The aim of this work is to characterize the binding profile of this opioid ligand on two duplicate delta receptors from zebrafish, ZFOR1 and ZFOR4. Our results show that, while ZFOR1 presents one single binding site for [(3)H]-MEGY (K(D)=4.0+/-0.4 nM), the experimental data from ZFOR4 fit better to the two-site binding model (K(D1)=0.8+/-0.2 nM and K(D2)=30.2+/-10.2 nM). Two other MEGY synthetic analogues, (D-Ala(2))-MEGY and (D-Ala(2), Val(5))-MEGY were also prepared and tested, together with the original peptide MEGY and other opioid ligands, in competition binding assays. While these peptides presented K(i) values on the nanomolar range when using [(3)H]-MEGY as radioligand, these parameters were two orders higher in competition binding assays with the antagonist [(3)H]-diprenorphine. Functional [(35)S]GTPgammaS stimulation analysis has revealed that these two receptors can be activated by several opioid agonists. Our results prove that although the MEGY peptide acts as an agonist on ZFOR1 and ZFOR4, there are subtle pharmacological differences between these two delta opioid receptors from zebrafish.


Asunto(s)
Encefalina Metionina/análogos & derivados , Receptores Opioides delta/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Unión Competitiva , Línea Celular , Diprenorfina/metabolismo , Encefalina Metionina/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Técnicas In Vitro , Cinética , Ligandos , Unión Proteica , Ratas , Receptores Opioides delta/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/genética
12.
Brain Res ; 1184: 46-56, 2007 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-17980352

RESUMEN

Lipid rafts are small cholesterol- and glycosphingolipid-enriched membrane subdomains. Here we compared the mu opioid receptor (MOR)-lipid rafts relationship in the rat brain, where neurons have non-caveolae rafts, and in CHO cells stably transfected with HA-rat MOR (CHO-HA-rMOR), which are enriched in caveolae. Membranes of rat caudate putamen (CPu) and thalamus or CHO-HA-rMOR cells were homogenized, sonicated in a detergent-free 0.5 M Na(2)CO(3) buffer and fractionated through sucrose density gradients. Western blot and [(3)H]diprenorphine binding showed that approximately 70% of MOR in CHO-HA-rMOR was present in low-density (5-20% sucrose) fractions enriched in cholesterol and/or ganglioside M1 (GM1) (lipid rafts) in plasma membranes, whereas about 70% and 45% of MOR in CPu and thalamus, respectively, were associated with lipid rafts. Incubation with a saturating concentration of etorphine or morphine at 37 degrees C for 30 min failed to change the MOR location in rafts in CHO-HA-rMOR, indicating that the internalized MOR does not move out of rafts, in contrast to the delta opioid receptor. In vivo, rafts association of MOR in CPu and thalamus was not affected significantly in rats implanted with two 75-mg morphine pellets for 72 h. In addition, cholesterol reduction by methyl-beta-cyclodextrin (MCD) disrupted rafts and shifted MOR to higher density fractions in both CHO-HA-rMOR and CPu membranes. However, MCD treatment had opposite impacts on MOR signaling in the two tissues: it attenuated MOR-mediated [(35)S]GTPgammaS binding in CPu but enhanced it in CHO-HA-rMOR.


Asunto(s)
Encéfalo/ultraestructura , Colesterol/metabolismo , Microdominios de Membrana/metabolismo , Receptores Opioides mu/metabolismo , Transducción de Señal/fisiología , Animales , Unión Competitiva/efectos de los fármacos , Células CHO/efectos de los fármacos , Células CHO/ultraestructura , Caveolina 1/metabolismo , Cricetinae , Cricetulus , Diprenorfina/metabolismo , Relación Dosis-Respuesta a Droga , Etorfina/farmacología , Masculino , Microdominios de Membrana/efectos de los fármacos , Morfina/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos/metabolismo , Antagonistas de Narcóticos/farmacología , Unión Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transfección
13.
J Mol Biol ; 429(12): 1840-1851, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28502792

RESUMEN

The µ-opioid receptor (MOPr) is a clinically important G protein-coupled receptor that couples to Gi/o proteins and arrestins. At present, the receptor conformational changes that occur following agonist binding and activation are poorly understood. This study has employed molecular dynamics simulations to investigate the binding mode and receptor conformational changes induced by structurally similar opioid ligands of widely differing intrinsic agonist efficacy, norbuprenorphine, buprenorphine, and diprenorphine. Bioluminescence resonance energy transfer assays for Gi activation and arrestin-3 recruitment in human embryonic kidney 293 cells confirmed that norbuprenorphine is a high efficacy agonist, buprenorphine a low efficacy agonist, and diprenorphine an antagonist at the MOPr. Molecular dynamics simulations revealed that these ligands adopt distinct binding poses and engage different subsets of residues, despite sharing a common morphinan scaffold. Notably, norbuprenorphine interacted with sodium ion-coordinating residues W2936.48 and N1503.35, whilst buprenorphine and diprenorphine did not. Principal component analysis of the movements of the receptor transmembrane domains showed that the buprenorphine-bound receptor occupied a distinct set of conformations to the norbuprenorphine-bound receptor. Addition of an allosteric sodium ion caused the receptor and ligand to adopt an inactive conformation. The differences in ligand-residue interactions and receptor conformations observed here may underlie the differing efficacies for cellular signalling outputs for these ligands.


Asunto(s)
Narcóticos/química , Narcóticos/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Buprenorfina/análogos & derivados , Buprenorfina/química , Buprenorfina/metabolismo , Línea Celular , Diprenorfina/química , Diprenorfina/metabolismo , Humanos , Mediciones Luminiscentes , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica
14.
Biochim Biophys Acta ; 1135(2): 226-8, 1992 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-1616942

RESUMEN

The study of the kinetics of binding of the opiate receptor agonist [3H]DADLE with NG108-15 cell suspensions has revealed a new periodic biological phenomenon, i.e., oscillations of the cellular receptor activity. The absence of oscillations for binding of the receptor antagonist shows that oscillations occur as a result of the transformation of the receptor signal only.


Asunto(s)
Membrana Celular/metabolismo , Diprenorfina/metabolismo , Leucina Encefalina-2-Alanina/metabolismo , Receptores de Droga/metabolismo , Animales , Línea Celular , Cinética , Sensibilidad y Especificidad
15.
Biochim Biophys Acta ; 685(3): 241-8, 1982 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-6279149

RESUMEN

(1) As previously shown, stereospecific binding of opiates to membrane bound receptors is inhibited by treatment with small amounts of phospholipase A2 from Vipera russelli. This effect is quantified and compared with the enzymes from the venoms of Naja Naja siamensis, Apis Mellifica and from porcine pancreas. All enzymes are equally effective. The inhibition is due to partial phospholipid hydrolysis leading to inactivation of membrane-bound receptor. (2) Bee venom phospholipase A2 together with the synergistically acting peptide, melittin, causes receptor solubilization up to 80% of preformed receptor-ligand complex can be solubilized in this manner. (3) Lysophosphatidylcholine, a product of phospholipid hydrolysis, solubilizes performed receptor-ligand complex to a similar extent. Several other detergents were tested for their ability to solubilize receptor-ligand complex. Digitonin appears to be most effective in solubilizing such a complex.


Asunto(s)
Encéfalo/metabolismo , Fosfolipasas A/farmacología , Fosfolipasas/farmacología , Receptores Opioides/metabolismo , Animales , Membrana Celular/metabolismo , Detergentes , Diprenorfina/metabolismo , Etorfina/metabolismo , Cinética , Fosfolipasas A2 , Ratas , Receptores Opioides/efectos de los fármacos , Receptores Opioides/aislamiento & purificación , Solubilidad
16.
Curr Top Med Chem ; 5(3): 303-13, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15857313

RESUMEN

Arylacetamide analgesics that stimulate kappa opioid receptors in the central nervous system mediate dysphoria and psychosis as well as analgesia. However, the naturally occurring peptide agonist, dynorphin A, is analgesic in the absence of dysphoria and psychosis, indicating that the therapeutic effects of kappa opioid agonists may be separated from their side effects. As part of our effort to discover kappa opioid receptor analgesics lacking side effects, we designed and constructed two mu/kappa chimeric receptors, composed primarily of amino acid residues derived from the mu opioid receptor, that were expected to bind dynorphin A with high affinity. In one, extracellular loop 2 and transmembrane domain 4 were derived from the kappa opioid receptor and in the other, only extracellular loop 2 was derived from the kappa opioid receptor. Most competitors of [(3)H]diprenorphine binding from a variety of structural classes bound to the chimeras with affinities similar to those with which they bound to the mu opioid receptor. In contrast, dynorphin A analogs bound to the chimeras with affinities similar to those with which they bound to the kappa opioid receptor. Pharmacological characterization of [(35)S]GTPgammaS binding mediated by the chimera with extracellular loop 2 derived from the kappa opioid receptor showed that it behaved as if it were mu opioid receptor with high affinity for dynorphin A analogs. These chimeras may be useful in identifying novel kappa receptor agonists that bind to the second extracellular loop of the receptor and share the desirable therapeutic profile of dynorphin A.


Asunto(s)
Dinorfinas/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Secuencia de Aminoácidos , Unión Competitiva/efectos de los fármacos , Diprenorfina/metabolismo , Diprenorfina/farmacología , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Humanos , Datos de Secuencia Molecular , Receptores Opioides kappa/genética , Receptores Opioides mu/genética
17.
J Mol Neurosci ; 27(2): 185-94, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16186629

RESUMEN

Arachidonic acid (AA), released in response to muscarinic acetylcholine receptor (mAChR) stimulation, previously has been reported to function as a reversible feedback inhibitor of the mAChR. To determine if the effects of AA on binding to the mAChR are subtype specific and whether AA inhibits ligand binding to other G protein-coupled receptors (GPCRs), the effects of AA on ligand binding to the mAChR subtypes (M1, M2, M3, M4, and M5) and to the micro-opioid receptor, beta2-adrenergic receptor (beta2-AR), 5-hydroxytryptamine receptor (5-HTR), and nicotinic receptors were examined. AA was found to inhibit ligand binding to all mAChR subtypes, to the beta2-AR, the 5-HTR, and to the micro-opioid receptor. However, AA does not inhibit ligand binding to the nicotinic receptor, even at high concentrations of AA. Thus, AA inhibits several types of GPCRs, with 50% inhibition occurring at 3-25 MuM, whereas the nicotinic receptor, a non-GPCR, remains unaffected. Further research is needed to determine the mechanism by which AA inhibits GPCR function.


Asunto(s)
Ácido Araquidónico/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Muscarínicos/metabolismo , Receptores Opioides mu/metabolismo , Receptores de Serotonina/metabolismo , Antagonistas Adrenérgicos beta/química , Antagonistas Adrenérgicos beta/metabolismo , Animales , Ácido Araquidónico/química , Dihidroalprenolol/química , Dihidroalprenolol/metabolismo , Diprenorfina/química , Diprenorfina/metabolismo , Humanos , Ligandos , Estructura Molecular , Antagonistas Muscarínicos/química , Antagonistas Muscarínicos/metabolismo , N-Metilescopolamina/química , N-Metilescopolamina/metabolismo , Antagonistas de Narcóticos/química , Antagonistas de Narcóticos/metabolismo , Parasimpatolíticos/química , Parasimpatolíticos/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Quinuclidinil Bencilato/química , Quinuclidinil Bencilato/metabolismo , Ensayo de Unión Radioligante , Serotonina/química , Serotonina/metabolismo
18.
J Cereb Blood Flow Metab ; 11(1): 1-9, 1991 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1845764

RESUMEN

The regional binding of the opiate receptor ligand diprenorphine has been examined in rat brain both in vivo and in vitro. The time course of total label in specific brain regions was followed up to 2 h after intravenous bolus injection of [3H]diprenorphine, with or without a pulse chase of unlabelled diprenorphine at 30 min. In addition, total label was measured 30 min after injection of labelled diprenorphine at nontracer concentrations over a range of specific activities. Total data sets for each region were fitted simultaneously to a compartmental model to give estimates of maximal binding capacity (Bmax), the second-order apparent association rate constant, and the first-order dissociation rate constant of the receptor-ligand complex. The model incorporated the use of a reference region with low specific binding (cerebellum). The binding of diprenorphine to rat brain homogenates was measured in vitro under equilibrium conditions at 37 degrees C, pH 7.4, in the presence and absence of naloxone, to give corresponding regional estimates of Bmax and the half-saturation constant Kd. The results showed a close correlation between in vitro and in vivo regional estimates of Bmax over a wide range. There were no significant interregional differences either in Kd in vitro or in the Kd derived from the in vivo analysis, although in vitro and in vivo estimates differed by an order of magnitude. This work was carried out as part of a validation study with a view to the application of the compartmental model to data obtained in vivo in humans using positron emission tomography, when successive studies over a range of specific activities are not feasible.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Encéfalo/metabolismo , Diprenorfina/metabolismo , Receptores Opioides/metabolismo , Animales , Cerebelo/metabolismo , Diprenorfina/farmacocinética , Cinética , Masculino , Ratas , Ratas Endogámicas , Tálamo/metabolismo , Distribución Tisular
19.
J Cereb Blood Flow Metab ; 19(7): 803-8, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10413036

RESUMEN

The binding of [11C]diprenorphine to mu, kappa, and delta subsites in cortical and subcortical structures was measured by positron emission tomography in vivo in six patients before and after surgical relief of trigeminal neuralgia pain. The volume of distribution of [11C]diprenorphine binding was significantly increased after thermocoagulation of the relevant trigeminal division in the following areas: prefrontal, insular, perigenual, mid-cingulate and inferior parietal cortices, basal ganglia, and thalamus bilaterally. In addition to the pain relief associated with the surgical procedure, there also was an improvement in anxiety and depression scores. In the context of other studies, these changes in binding most likely resulted from the change in the pain state. The results suggest an increased occupancy by endogenous opioid peptides during trigeminal pain but cannot exclude coexistent down-regulation of binding sites.


Asunto(s)
Diprenorfina/metabolismo , Receptores Opioides/fisiología , Neuralgia del Trigémino/fisiopatología , Anciano , Radioisótopos de Carbono , Humanos , Masculino , Persona de Mediana Edad , Antagonistas de Narcóticos/metabolismo , Ensayo de Unión Radioligante , Tomografía Computarizada de Emisión , Neuralgia del Trigémino/metabolismo
20.
J Cereb Blood Flow Metab ; 10(4): 484-92, 1990 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2161414

RESUMEN

The kinetics and regional distribution of [11C]carfentanil, a mu-selective opiate receptor agonist, and [11C]diprenorphine, a nonselective opiate receptor antagonist, were compared using paired positron emission tomography studies in two normal volunteers. Kinetics of total radioactivity (counts/mCi/pixel) was greater for [11C]diprenorphine than [11C]carfentanil in all regions. [11C]Carfentanil binding (expressed as the total/nonspecific ratio) reached near equilibrium at approximately 40 min, whereas [11C]diprenorphine showed a linear increase until approximately 60 min. Kinetics of specific binding demonstrated significant dissociation of [11C]carfentanil from opiate receptors, whereas little dissociation of [11C]diprenorphine was observed during the 90-min scan session. Regional distributions of [11C]carfentanil and [11C]diprenorphine were qualitatively and quantitatively different: Relative to the thalamus (a region with known predominance of mu-receptors), [11C]diprenorphine displayed greater binding in the striatum and cingulate and frontal cortex compared to [11C]carfentanil, consistent with labeling of additional, non-mu sites by [11C]diprenorphine. We conclude from these studies that [11C]diprenorphine labels other opiate receptor subtypes in addition to the mu sites selectively labeled by [11C]carfentanil. The nonselective nature of diprenorphine potentially limits its usefulness in defining abnormalities of specific opiate receptor subtypes in various diseases. Development of selective tracers for the delta- and kappa-opiate receptor sites, or alternatively use of unlabeled inhibitors to differentially displace mu, delta, and kappa subtypes, will help offset these limitations.


Asunto(s)
Encéfalo/metabolismo , Diprenorfina/metabolismo , Fentanilo/análogos & derivados , Morfinanos/metabolismo , Receptores Opioides/metabolismo , Tomografía Computarizada de Emisión , Adulto , Encéfalo/diagnóstico por imagen , Fentanilo/metabolismo , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA